101
|
McMenamin AJ, Daughenbaugh KF, Flenniken ML. The Heat Shock Response in the Western Honey Bee (Apis mellifera) is Antiviral. Viruses 2020; 12:E245. [PMID: 32098425 PMCID: PMC7077298 DOI: 10.3390/v12020245] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/22/2022] Open
Abstract
Honey bees (Apismellifera) are an agriculturally important pollinator species that live in easily managed social groups (i.e., colonies). Unfortunately, annual losses of honey bee colonies in many parts of the world have reached unsustainable levels. Multiple abiotic and biotic stressors, including viruses, are associated with individual honey bee and colony mortality. Honey bees have evolved several antiviral defense mechanisms including conserved immune pathways (e.g., Toll, Imd, JAK/STAT) and dsRNA-triggered responses including RNA interference and a non-sequence specific dsRNA-mediated response. In addition, transcriptome analyses of virus-infected honey bees implicate an antiviral role of stress response pathways, including the heat shock response. Herein, we demonstrate that the heat shock response is antiviral in honey bees. Specifically, heat-shocked honey bees (i.e., 42 °C for 4 h) had reduced levels of the model virus, Sindbis-GFP, compared with bees maintained at a constant temperature. Virus-infection and/or heat shock resulted in differential expression of six heat shock protein encoding genes and three immune genes, many of which are positively correlated. The heat shock protein encoding and immune gene transcriptional responses observed in virus-infected bees were not completely recapitulated by administration of double stranded RNA (dsRNA), a virus-associated molecular pattern, indicating that additional virus-host interactions are involved in triggering antiviral stress response pathways.
Collapse
Affiliation(s)
- Alexander J. McMenamin
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT 59717, USA; (A.J.M.); (K.F.D.)
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
- Pollinator Health Center, Montana State University, Bozeman, MT 59717, USA
| | - Katie F. Daughenbaugh
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT 59717, USA; (A.J.M.); (K.F.D.)
- Pollinator Health Center, Montana State University, Bozeman, MT 59717, USA
| | - Michelle L. Flenniken
- Department of Plant Sciences and Plant Pathology, Montana State University, Bozeman, MT 59717, USA; (A.J.M.); (K.F.D.)
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
- Pollinator Health Center, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
102
|
Martin M, Hiroyasu A, Guzman RM, Roberts SA, Goodman AG. Analysis of Drosophila STING Reveals an Evolutionarily Conserved Antimicrobial Function. Cell Rep 2019; 23:3537-3550.e6. [PMID: 29924997 DOI: 10.1016/j.celrep.2018.05.029] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 03/15/2018] [Accepted: 05/08/2018] [Indexed: 01/24/2023] Open
Abstract
The vertebrate protein STING, an intracellular sensor of cyclic dinucleotides, is critical to the innate immune response and the induction of type I interferon during pathogenic infection. Here, we show that a STING ortholog (dmSTING) exists in Drosophila, which, similar to vertebrate STING, associates with cyclic dinucleotides to initiate an innate immune response. Following infection with Listeria monocytogenes, dmSTING activates an innate immune response via activation of the NF-κB transcription factor Relish, part of the immune deficiency (IMD) pathway. DmSTING-mediated activation of the immune response reduces the levels of Listeria-induced lethality and bacterial load in the host. Of significance, dmSTING triggers an innate immune response in the absence of a known functional cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) ortholog in the fly. Together, our results demonstrate that STING is an evolutionarily conserved antimicrobial effector between flies and mammals, and it comprises a key component of host defense against pathogenic infection in Drosophila.
Collapse
Affiliation(s)
- Marina Martin
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Aoi Hiroyasu
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - R Marena Guzman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Steven A Roberts
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA; Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA.
| |
Collapse
|
103
|
Drosophila immunity against natural and nonnatural viral pathogens. Virology 2019; 540:165-171. [PMID: 31928998 DOI: 10.1016/j.virol.2019.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/01/2019] [Indexed: 01/25/2023]
Abstract
The fruit fly Drosophila melanogaster is extensively used as a model species for molecular biology and genetics. It is also widely studied for its innate immune system to expand our understanding of immune host defenses against numerous pathogens. More precisely, studies using both natural and nonnatural Drosophila pathogens have provided a better perspective of viral infection strategies and immunity processes than any other invertebrate. This has made significant advances in identifying and characterizing the innate immune mechanisms by which hosts can combat viral pathogens. However, in-depth studies on antiviral immunity are still lacking due in part to the narrow research focus on the evolution and conservation of antiviral strategies to combat infections caused by both natural and nonnatural viruses. In this review, we will cover three major areas. First, we will describe the well-characterized antiviral immune mechanisms in Drosophila. Second, we will survey the specific pathways induced by natural viruses that have been studied in Drosophila. Finally, we will discuss the pathways activated by nonnatural viruses, drawing comparisons to natural viruses and giving an unprecedented insight into the virus community of Drosophila that is necessary to understand the evolutionary and immune context needed to develop Drosophila as a model for virus research.
Collapse
|
104
|
Blair CD. Deducing the Role of Virus Genome-Derived PIWI-Associated RNAs in the Mosquito-Arbovirus Arms Race. Front Genet 2019; 10:1114. [PMID: 31850054 PMCID: PMC6901949 DOI: 10.3389/fgene.2019.01114] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/16/2019] [Indexed: 01/26/2023] Open
Abstract
The P-element-induced wimpy testis (PIWI)-associated RNA (piRNA) pathway is known for its role in the protection of genome integrity in the germline of Drosophila melanogaster by silencing transposable elements. The piRNAs that target transposons originate from piRNA clusters in transposon-rich regions of the Drosophila genome and are processed by three PIWI family proteins. In Aedes aegypti and Aedes albopictus mosquitoes, which are two of the most important vectors of arthropod-borne viruses (arboviruses), the number of PIWI family genes has expanded and some are expressed in somatic, as well as germline, tissues. These discoveries have led to active research to explore the possible expanded functional roles of the piRNA pathway in vector mosquitoes. Virus genome-derived piRNAs (which will be referred to as (virus name) vpiRNAs) have been demonstrated in Aedes spp. cultured cells and mosquitoes after infection by arthropod-borne alpha-, bunya-, and flaviviruses. However, although Culex quinquefasciatus also is an important arbovirus vector and has an expansion of PIWI family genes, vpiRNAs have seldom been documented in this genus after arbovirus infection. Generation of complementary DNA (cDNA) fragments from RNA genomes of alpha-, bunya-, and flaviviruses (viral-derived cDNAs, vDNAs) has been demonstrated in cultured Aedes spp. cells and mosquitoes, and endogenous viral elements (EVEs), cDNA fragments of non-retroviral RNA virus genomes, are found more abundantly in genomes of Ae. aegypti and Ae. albopictus than other vector mosquitoes. These observations have led to speculation that vDNAs are integrated into vector genomes to form EVEs, which serve as templates for the transcription of antiviral vpiRNA precursors. However, no EVEs derived from alphavirus genomes have been demonstrated in genomes of any vector mosquito. In addition, although EVEs have been shown to be a source of piRNAs, the preponderance of EVEs described in Aedes spp. vectors are more closely related to the genomes of persistently infecting insect-specific viruses than to acutely infecting arboviruses. Furthermore, the signature patterns of the “ping-pong” amplification cycle that maintains transposon-targeting piRNAs in Drosophila are also evident in alphavirus and bunyavirus vpiRNAs, but not in vpiRNAs of flaviviruses. These divergent observations have rendered deciphering the mechanism(s) of biogenesis and potential role of vpiRNAs in the mosquito–arbovirus arms race difficult, and the focus of this review will be to assemble major findings regarding vpiRNAs and antiviral immunity in the important arbovirus vectors from Aedes and Culex genera.
Collapse
Affiliation(s)
- Carol D Blair
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
105
|
A DNA virus-encoded immune antagonist fully masks the potent antiviral activity of RNAi in Drosophila. Proc Natl Acad Sci U S A 2019; 116:24296-24302. [PMID: 31712431 DOI: 10.1073/pnas.1909183116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Coevolution of viruses and their hosts may lead to viral strategies to avoid, evade, or suppress antiviral immunity. An example is antiviral RNA interference (RNAi) in insects: the host RNAi machinery processes viral double-stranded RNA into small interfering RNAs (siRNAs) to suppress viral replication, whereas insect viruses encode suppressors of RNAi, many of which inhibit viral small interfering RNA (vsiRNA) production. Yet, many studies have analyzed viral RNAi suppressors in heterologous systems, due to the lack of experimental systems to manipulate the viral genome of interest, raising questions about in vivo functions of RNAi suppressors. To address this caveat, we generated an RNAi suppressor-defective mutant of invertebrate iridescent virus 6 (IIV6), a large DNA virus in which we previously identified the 340R protein as a suppressor of RNAi. Loss of 340R did not affect vsiRNA production, indicating that 340R binds siRNA duplexes to prevent RNA-induced silencing complex assembly. Indeed, vsiRNAs were not efficiently loaded into Argonaute 2 during wild-type IIV6 infection. Moreover, IIV6 induced a limited set of mature microRNAs in a 340R-dependent manner, most notably miR-305-3p, which we attribute to stabilization of the miR-305-5p:3p duplex by 340R. The IIV6 340R deletion mutant did not have a replication defect in cells, but was strongly attenuated in adult Drosophila This in vivo replication defect was completely rescued in RNAi mutant flies, indicating that 340R is a bona fide RNAi suppressor, the absence of which uncovers a potent antiviral immune response that suppresses virus accumulation ∼100-fold. Together, our work indicates that viral RNAi suppressors may completely mask antiviral immunity.
Collapse
|
106
|
Fredericks AC, Russell TA, Wallace LE, Davidson AD, Fernandez-Sesma A, Maringer K. Aedes aegypti (Aag2)-derived clonal mosquito cell lines reveal the effects of pre-existing persistent infection with the insect-specific bunyavirus Phasi Charoen-like virus on arbovirus replication. PLoS Negl Trop Dis 2019; 13:e0007346. [PMID: 31693659 DOI: 10.1101/596205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/18/2019] [Accepted: 10/24/2019] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Aedes aegypti is a vector mosquito of major public health importance, transmitting arthropod-borne viruses (arboviruses) such as chikungunya, dengue, yellow fever and Zika viruses. Wild mosquito populations are persistently infected at high prevalence with insect-specific viruses that do not replicate in vertebrate hosts. In experimental settings, acute infections with insect-specific viruses have been shown to modulate arbovirus infection and transmission in Ae. aegypti and other vector mosquitoes. However, the impact of persistent insect-specific virus infections, which arboviruses encounter more commonly in nature, has not been investigated extensively. Cell lines are useful models for studying virus-host interactions, however the available Ae. aegypti cell lines are poorly defined and heterogenous cultures. METHODOLOGY/PRINCIPLE FINDINGS We generated single cell-derived clonal cell lines from the commonly used Ae. aegypti cell line Aag2. Two of the fourteen Aag2-derived clonal cell lines generated harboured markedly and consistently reduced levels of the insect-specific bunyavirus Phasi Charoen-like virus (PCLV) known to persistently infect Aag2 cells. In contrast to studies with acute insect-specific virus infections in cell culture and in vivo, we found that pre-existing persistent PCLV infection had no major impact on the replication of the flaviviruses dengue virus and Zika virus, the alphavirus Sindbis virus, or the rhabdovirus vesicular stomatitis virus. We also performed a detailed characterisation of the morphology, transfection efficiency and immune status of our Aag2-derived clonal cell lines, and have made a clone that we term Aag2-AF5 available to the research community as a well-defined cell culture model for arbovirus-vector interaction studies. CONCLUSIONS/SIGNIFICANCE Our findings highlight the need for further in vivo studies that more closely recapitulate natural arbovirus transmission settings in which arboviruses encounter mosquitoes harbouring persistent rather than acute insect-specific virus infections. Furthermore, we provide the well-characterised Aag2-derived clonal cell line as a valuable resource to the arbovirus research community.
Collapse
Affiliation(s)
- Anthony C Fredericks
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Tiffany A Russell
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Louisa E Wallace
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Andrew D Davidson
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kevin Maringer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Microbial Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
107
|
Tassetto M, Kunitomi M, Whitfield ZJ, Dolan PT, Sánchez-Vargas I, Garcia-Knight M, Ribiero I, Chen T, Olson KE, Andino R. Control of RNA viruses in mosquito cells through the acquisition of vDNA and endogenous viral elements. eLife 2019; 8:41244. [PMID: 31621580 PMCID: PMC6797480 DOI: 10.7554/elife.41244] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 09/10/2019] [Indexed: 01/02/2023] Open
Abstract
Aedes aegypti transmit pathogenic arboviruses while the mosquito itself tolerates the infection. We examine a piRNA-based immunity that relies on the acquisition of viral derived cDNA (vDNA) and how this pathway discriminates between self and non-self. The piRNAs derived from these vDNAs are essential for virus control and Piwi4 has a central role in the pathway. Piwi4 binds preferentially to virus-derived piRNAs but not to transposon-targeting piRNAs. Analysis of episomal vDNA from infected cells reveals that vDNA molecules are acquired through a discriminatory process of reverse-transcription and recombination directed by endogenous retrotransposons. Using a high-resolution Ae. aegypti genomic sequence, we found that vDNAs integrated in the host genome as endogenous viral elements (EVEs), produce antisense piRNAs that are preferentially loaded onto Piwi4. Importantly, EVE-derived piRNAs are specifically loaded onto Piwi4 to inhibit virus replication. Thus, Ae. aegypti employs a sophisticated antiviral mechanism that promotes viral persistence and generates long-lasting adaptive immunity.
Collapse
Affiliation(s)
- Michel Tassetto
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Mark Kunitomi
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Zachary J Whitfield
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Patrick T Dolan
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Irma Sánchez-Vargas
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, United States
| | - Miguel Garcia-Knight
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Isabel Ribiero
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Taotao Chen
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| | - Ken E Olson
- Department of Microbiology, Immunology and Pathology, Arthropod-borne and Infectious Diseases Laboratory, Colorado State University, Fort Collins, United States
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
108
|
Agboli E, Leggewie M, Altinli M, Schnettler E. Mosquito-Specific Viruses-Transmission and Interaction. Viruses 2019; 11:v11090873. [PMID: 31533367 PMCID: PMC6784079 DOI: 10.3390/v11090873] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Mosquito-specific viruses (MSVs) are a subset of insect-specific viruses that are found to infect mosquitoes or mosquito derived cells. There has been an increase in discoveries of novel MSVs in recent years. This has expanded our understanding of viral diversity and evolution but has also sparked questions concerning the transmission of these viruses and interactions with their hosts and its microbiome. In fact, there is already evidence that MSVs interact with the immune system of their host. This is especially interesting, since mosquitoes can be infected with both MSVs and arthropod-borne (arbo) viruses of public health concern. In this review, we give an update on the different MSVs discovered so far and describe current data on their transmission and interaction with the mosquito immune system as well as the effect MSVs could have on an arboviruses-co-infection. Lastly, we discuss potential uses of these viruses, including vector and transmission control.
Collapse
Affiliation(s)
- Eric Agboli
- Molecular Entomology, Molecular Biology and Immunology Department, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
- Department of Epidemiology and Biostatistics, School of Public Health, University of Health and Allied Sciences, Ho PMB 31, Ghana.
| | - Mayke Leggewie
- Molecular Entomology, Molecular Biology and Immunology Department, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
- German Centre for Infection research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany.
| | - Mine Altinli
- Molecular Entomology, Molecular Biology and Immunology Department, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
- German Centre for Infection research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany.
| | - Esther Schnettler
- Molecular Entomology, Molecular Biology and Immunology Department, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany.
- German Centre for Infection research (DZIF), partner site Hamburg-Lübeck-Borstel-Riems, 20359 Hamburg, Germany.
| |
Collapse
|
109
|
Karamipour N, Fathipour Y, Talebi AA, Asgari S, Mehrabadi M. The microRNA pathway is involved in Spodoptera frugiperda (Sf9) cells antiviral immune defense against Autographa californica multiple nucleopolyhedrovirus infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 112:103202. [PMID: 31422153 DOI: 10.1016/j.ibmb.2019.103202] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
The microRNA (miRNA) pathway is an epigenetic mechanism that plays important roles in various biological processes including host-virus interactions by regulating gene expression of the host and/or the virus. Previously, we showed that the cellular microRNAome in Spodoptera frugiperda (Sf9) cells is modulated following Autographa californica multiple nucleopolyhedrovirus (AcMNPV) infection suggesting that miRNAs may contribute in the cellular antiviral immunity. Here, we investigated the role of core components of the miRNA pathway in Sf9-AcMNPV interaction. Gene expression analyses showed that the expression levels of Dicer-1 (Dcr1), Argonaute-1 (Ago1) and Exportin-5 (Exp5) increased following AcMNPV infection particularly at 16 h post infection (hpi). Ran expression levels, however, decreased in response to virus infection. The expression levels of cellular miRNAs, miR-184 and let-7, also diminished at the post infection times further confirming differential expression of the cellular miRNAs following AcMNPV infection. To determine the role of the miRNA pathway in the interaction, we silenced key genes in the pathway using specific dsRNAs. RNAi of Dcr1, Ago1 and Ran enhanced viral DNA replication and reduced the abundance of miR-184 and let-7 underscoring the importance of the miRNA pathway in antiviral immunity in Sf9 cells. Suppression of the miRNA pathway in mock and infected cells had no effect on Ran expression levels suggesting miRNA-independent downregulation of this gene after virus infection. In conclusion, our results suggest the antiviral role of the miRNA pathway in Sf9 cells against AcMNPV. To modulate this immune response, AcMNPV represses host miRNAs likely through downregulation of Ran to enhance its replication in the host cells.
Collapse
Affiliation(s)
- Naeime Karamipour
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathipour
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Asghar Talebi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Sassan Asgari
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Mohammad Mehrabadi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
110
|
Discovery and characterization of a novel alphavirus-like RNA virus from the red firebug Pyrrhocoris apterus L. (Heteroptera). J Invertebr Pathol 2019; 166:107213. [DOI: 10.1016/j.jip.2019.107213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 11/22/2022]
|
111
|
Majzoub K, Wrensch F, Baumert TF. The Innate Antiviral Response in Animals: An Evolutionary Perspective from Flagellates to Humans. Viruses 2019; 11:v11080758. [PMID: 31426357 PMCID: PMC6723221 DOI: 10.3390/v11080758] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
Animal cells have evolved dedicated molecular systems for sensing and delivering a coordinated response to viral threats. Our understanding of these pathways is almost entirely defined by studies in humans or model organisms like mice, fruit flies and worms. However, new genomic and functional data from organisms such as sponges, anemones and mollusks are helping redefine our understanding of these immune systems and their evolution. In this review, we will discuss our current knowledge of the innate immune pathways involved in sensing, signaling and inducing genes to counter viral infections in vertebrate animals. We will then focus on some central conserved players of this response including Toll-like receptors (TLRs), RIG-I-like receptors (RLRs) and cGAS-STING, attempting to put their evolution into perspective. To conclude, we will reflect on the arms race that exists between viruses and their animal hosts, illustrated by the dynamic evolution and diversification of innate immune pathways. These concepts are not only important to understand virus-host interactions in general but may also be relevant for the development of novel curative approaches against human disease.
Collapse
Affiliation(s)
- Karim Majzoub
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 67000 Strasbourg, France.
| | - Florian Wrensch
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 67000 Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, 67000 Strasbourg, France.
- Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France.
- Institut Universitaire de France, 75231 Paris, France.
| |
Collapse
|
112
|
Pajak A, Laine I, Clemente P, El-Fissi N, Schober FA, Maffezzini C, Calvo-Garrido J, Wibom R, Filograna R, Dhir A, Wedell A, Freyer C, Wredenberg A. Defects of mitochondrial RNA turnover lead to the accumulation of double-stranded RNA in vivo. PLoS Genet 2019; 15:e1008240. [PMID: 31365523 PMCID: PMC6668790 DOI: 10.1371/journal.pgen.1008240] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/11/2019] [Indexed: 12/02/2022] Open
Abstract
The RNA helicase SUV3 and the polynucleotide phosphorylase PNPase are involved in the degradation of mitochondrial mRNAs but their roles in vivo are not fully understood. Additionally, upstream processes, such as transcript maturation, have been linked to some of these factors, suggesting either dual roles or tightly interconnected mechanisms of mitochondrial RNA metabolism. To get a better understanding of the turn-over of mitochondrial RNAs in vivo, we manipulated the mitochondrial mRNA degrading complex in Drosophila melanogaster models and studied the molecular consequences. Additionally, we investigated if and how these factors interact with the mitochondrial poly(A) polymerase, MTPAP, as well as with the mitochondrial mRNA stabilising factor, LRPPRC. Our results demonstrate a tight interdependency of mitochondrial mRNA stability, polyadenylation and the removal of antisense RNA. Furthermore, disruption of degradation, as well as polyadenylation, leads to the accumulation of double-stranded RNAs, and their escape out into the cytoplasm is associated with an altered immune-response in flies. Together our results suggest a highly organised and inter-dependable regulation of mitochondrial RNA metabolism with far reaching consequences on cellular physiology. Although a number of factors have been implemented in the turnover of mitochondrial (mt) DNA-derived transcripts, their exact functions and interplay with one another is not entirely clear. Several of these factors have been proposed to co-ordinately regulate both transcript maturation, as well as degradation, but the order of events during mitochondrial RNA turnover is less well understood. Using a range of different genetically modified Drosophila melanogaster models, we studied the involvement of the RNA helicase SUV3, the polynucleotide phosphorylase PNPase, the leucine-rich pentatricopeptide repeat motif-containing protein LRPPRC, and the mitochondrial RNA poly(A) polymerase MTPAP, in stabilisation, polyadenylation, and degradation of mitochondrial transcripts. Our results show a tight collaborative activity of these factors in vivo and reveal a clear hierarchical order of events leading to mitochondrial mRNA maturation. Furthermore, we demonstrate that the loss of SUV3, PNPase, or MTPAP leads to the accumulation of mitochondrial-derived antisense RNA in the cytoplasm of cells, which is associated with an altered immune-response in flies.
Collapse
Affiliation(s)
- Aleksandra Pajak
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Isabelle Laine
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Paula Clemente
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Najla El-Fissi
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Florian A. Schober
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Camilla Maffezzini
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Javier Calvo-Garrido
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Wibom
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Roberta Filograna
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Ashish Dhir
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Wedell
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Freyer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- * E-mail: (CF); (AW)
| | - Anna Wredenberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Max Planck Institute Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
- * E-mail: (CF); (AW)
| |
Collapse
|
113
|
Fareh M, van Lopik J, Katechis I, Bronkhorst AW, Haagsma AC, van Rij RP, Joo C. Viral suppressors of RNAi employ a rapid screening mode to discriminate viral RNA from cellular small RNA. Nucleic Acids Res 2019; 46:3187-3197. [PMID: 29325071 PMCID: PMC5888754 DOI: 10.1093/nar/gkx1316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/03/2018] [Indexed: 11/14/2022] Open
Abstract
RNA interference (RNAi) is an indispensable mechanism for antiviral defense in insects, including mosquitoes that transmit human diseases. To escape this antiviral defense system, viruses encode suppressors of RNAi that prevent elimination of viral RNAs, and thus ensure efficient virus accumulation. Although the first animal Viral Suppressor of RNAi (VSR) was identified more than a decade ago, the molecular basis of RNAi suppression by these viral proteins remains unclear. Here, we developed a single-molecule fluorescence assay to investigate how VSRs inhibit the recognition of viral RNAs by Dcr-2, a key endoribonuclease enzyme in the RNAi pathway. Using VSRs from three insect RNA viruses (Culex Y virus, Drosophila X virus and Drosophila C virus), we reveal bimodal physical interactions between RNA molecules and VSRs. During initial interactions, these VSRs rapidly discriminate short RNA substrates from long dsRNA. VSRs engage nearly irreversible binding with long dsRNAs, thereby shielding it from recognition by Dcr-2. We propose that the length-dependent switch from rapid screening to irreversible binding reflects the main mechanism by which VSRs distinguish viral dsRNA from cellular RNA species such as microRNAs.
Collapse
Affiliation(s)
- Mohamed Fareh
- Kavli Institute of NanoScience and Department of BioNanoScience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Jasper van Lopik
- Kavli Institute of NanoScience and Department of BioNanoScience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Iason Katechis
- Kavli Institute of NanoScience and Department of BioNanoScience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Alfred W Bronkhorst
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6525 GA, The Netherlands
| | - Anna C Haagsma
- Kavli Institute of NanoScience and Department of BioNanoScience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen 6525 GA, The Netherlands
| | - Chirlmin Joo
- Kavli Institute of NanoScience and Department of BioNanoScience, Delft University of Technology, Delft 2629 HZ, The Netherlands
| |
Collapse
|
114
|
Mallick B, Sharma AR, Lee SS, Chakraborty C. Understanding the molecular interaction of human argonaute-2 and miR-20a complex: A molecular dynamics approach. J Cell Biochem 2019; 120:19915-19924. [PMID: 31318096 DOI: 10.1002/jcb.29300] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 06/18/2019] [Indexed: 12/16/2022]
Abstract
Argonaute-2 (AGO2), a member of the Argonaute family, is the only member possessing catalytic and RNA silencing activity. In here, a molecular dynamics (MDs) simulation was performed using the crystal structure of human AGO2 protein complex with miR-20a. miR-20a is involved with various kind of biological process like heart and lung development, oncogenic process, etc. In precise, MD simulation was carried out with AGO2 protein complex with wild type, two mutant sites and four mutant sites in guided microRNA (miRNA). It has been noted that root-mean-square deviation (RMSD) of atomic positions of nucleic acid for wild type and two mutant sites guided miRNA has the same pattern of fluctuations, which stabilizes around 0.27 nm after 2 ns. Cα atom of AGO2 protein in the complex shows that this complex with wild type and two mutant site mutation duplex has a stable RMSD value after 20 ns, ranging between 0.14 and 0.21 nm. From the root-mean-square fluctuation (RMSF), we observed an increased pattern of fluctuations for the atoms of four mutant complex of AGO2-miR-20a complex. This increased RMSF of non-mutated nucleic acids is contributed by U-A bond breaking at the site of the nucleotide of U2 of guided miRNA, as observed from the duplex structure taken at different time steps of the simulation. Superimposed structure of the miRNA-mRNA duplex for the three complexes depicts that the three miRNA-mRNA duplexes are stable during the simulation. Current work demonstrates the possible correlations between the conformational changes of this AGO2-miR-20a duplex structure and the interactions of different atoms.
Collapse
Affiliation(s)
- Bidyut Mallick
- Departments of Physics, Galgotias College of Engineering and Technology, Greater Noida, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do, Republic of Korea
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do, Republic of Korea
| | - Chiranjib Chakraborty
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon, Gangwon-Do, Republic of Korea.,Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| |
Collapse
|
115
|
Antiviral RNAi in Insects and Mammals: Parallels and Differences. Viruses 2019; 11:v11050448. [PMID: 31100912 PMCID: PMC6563508 DOI: 10.3390/v11050448] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/26/2022] Open
Abstract
The RNA interference (RNAi) pathway is a potent antiviral defense mechanism in plants and invertebrates, in response to which viruses evolved suppressors of RNAi. In mammals, the first line of defense is mediated by the type I interferon system (IFN); however, the degree to which RNAi contributes to antiviral defense is still not completely understood. Recent work suggests that antiviral RNAi is active in undifferentiated stem cells and that antiviral RNAi can be uncovered in differentiated cells in which the IFN system is inactive or in infections with viruses lacking putative viral suppressors of RNAi. In this review, we describe the mechanism of RNAi and its antiviral functions in insects and mammals. We draw parallels and highlight differences between (antiviral) RNAi in these classes of animals and discuss open questions for future research.
Collapse
|
116
|
Eyk CLV, Samaraweera SE, Scott A, Webber DL, Harvey DP, Mecinger O, O’Keefe LV, Cropley JE, Young P, Ho J, Suter C, Richards RI. Non-self mutation: double-stranded RNA elicits antiviral pathogenic response in a Drosophila model of expanded CAG repeat neurodegenerative diseases. Hum Mol Genet 2019; 28:3000-3012. [DOI: 10.1093/hmg/ddz096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
Abstract
Inflammation is activated prior to symptoms in neurodegenerative diseases, providing a plausible pathogenic mechanism. Indeed, genetic and pharmacological ablation studies in animal models of several neurodegenerative diseases demonstrate that inflammation is required for pathology. However, while there is growing evidence that inflammation-mediated pathology may be the common mechanism underlying neurodegenerative diseases, including those due to dominantly inherited expanded repeats, the proximal causal agent is unknown. Expanded CAG.CUG repeat double-stranded RNA causes inflammation-mediated pathology when expressed in Drosophila. Repeat dsRNA is recognized by Dicer-2 as a foreign or ‘non-self’ molecule triggering both antiviral RNA and RNAi pathways. Neither of the RNAi pathway cofactors R2D2 nor loquacious are necessary, indicating antiviral RNA activation. RNA modification enables avoidance of recognition as ‘non-self’ by the innate inflammatory surveillance system. Human ADAR1 edits RNA conferring ‘self’ status and when co-expressed with expanded CAG.CUG dsRNA in Drosophila the pathology is lost. Cricket Paralysis Virus protein CrPV-1A is a known antagonist of Argonaute-2 in Drosophila antiviral defense. CrPV-1A co-expression also rescues pathogenesis, confirming anti-viral-RNA response. Repeat expansion mutation therefore confers ‘non-self’ recognition of endogenous RNA, thereby providing a proximal, autoinflammatory trigger for expanded repeat neurodegenerative diseases.
Collapse
Affiliation(s)
- Clare L van Eyk
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Saumya E Samaraweera
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Andrew Scott
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Dani L Webber
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - David P Harvey
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Olivia Mecinger
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Louise V O’Keefe
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Jennifer E Cropley
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Paul Young
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Joshua Ho
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Catherine Suter
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Robert I Richards
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
117
|
Maillard PV, van der Veen AG, Poirier EZ, Reis e Sousa C. Slicing and dicing viruses: antiviral RNA interference in mammals. EMBO J 2019; 38:e100941. [PMID: 30872283 PMCID: PMC6463209 DOI: 10.15252/embj.2018100941] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/17/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
To protect against the harmful consequences of viral infections, organisms are equipped with sophisticated antiviral mechanisms, including cell-intrinsic means to restrict viral replication and propagation. Plant and invertebrate cells utilise mostly RNA interference (RNAi), an RNA-based mechanism, for cell-intrinsic immunity to viruses while vertebrates rely on the protein-based interferon (IFN)-driven innate immune system for the same purpose. The RNAi machinery is conserved in vertebrate cells, yet whether antiviral RNAi is still active in mammals and functionally relevant to mammalian antiviral defence is intensely debated. Here, we discuss cellular and viral factors that impact on antiviral RNAi and the contexts in which this system might be at play in mammalian resistance to viral infection.
Collapse
Affiliation(s)
- Pierre V Maillard
- Division of Infection and Immunity, University College London, London, UK
| | | | - Enzo Z Poirier
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | |
Collapse
|
118
|
Barroso-Arévalo S, Vicente-Rubiano M, Puerta F, Molero F, Sánchez-Vizcaíno JM. Immune related genes as markers for monitoring health status of honey bee colonies. BMC Vet Res 2019; 15:72. [PMID: 30832657 PMCID: PMC6398266 DOI: 10.1186/s12917-019-1823-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 02/25/2019] [Indexed: 12/21/2022] Open
Abstract
Background Honey bee population decline threatens the beekeeping sector, agriculture and global biodiversity. Early detection of colony mortality may facilitate rapid interventions to contain and prevent mortality spread. Among others, deformed wing virus (DWV) is capable of inducing colony losses, especially when combined with Varroa destructor mite. Since the bee immune system plays a crucial role in ensuring that bees are able to face these pathogens, we explored whether expression of immune genes could serve as biomarkers of colony health. Results Herein, we describe a preliminary immunological marker composed of two immune genes (relish and defensin), which provide insight on honey bee antiviral defense mechanism. Of the tested genes, relish expression correlated with the presence of DWV-Varroa complex, while decreased defensin expression correlated with poor resistance to this complex. Conclusions The monitoring of these genes may help us to better understand the complex physiology of honey bees’s immune system and to develop new approaches for managing the health impacts of DWV infection and varroa infestation in the field.
Collapse
Affiliation(s)
- Sandra Barroso-Arévalo
- VISAVET Centre and Animal Health Department, Veterinary School, Complutense University of Madrid, Madrid, Spain.
| | - Marina Vicente-Rubiano
- VISAVET Centre and Animal Health Department, Veterinary School, Complutense University of Madrid, Madrid, Spain
| | - Francisco Puerta
- Apicultural Reference Center in Andalusia (CERA), Andalusia, Spain
| | - Fernando Molero
- Apicultural Reference Center in Andalusia (CERA), Andalusia, Spain
| | | |
Collapse
|
119
|
Marklewitz M, Junglen S. Evolutionary and ecological insights into the emergence of arthropod-borne viruses. Acta Trop 2019; 190:52-58. [PMID: 30339799 DOI: 10.1016/j.actatropica.2018.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/19/2018] [Accepted: 10/12/2018] [Indexed: 02/05/2023]
Abstract
The emergence of arthropod-borne viruses (arboviruses) is of global concern as they can rapidly spread across countries and to new continents as the recent examples of chikungunya virus and Zika virus have demonstrated. Whereas the global movement patterns of emerging arboviruses are comparatively well studied, there is little knowledge on initial emergence processes that enable sylvatic (enzootic) viruses to leave their natural amplification cycle and infect humans or livestock, often also involving infection of anthropophilic vector species. Emerging arboviruses almost exclusively originate in highly biodiverse ecosystems of tropical countries. Changes in host population diversity and density can affect pathogen transmission patterns and are likely to influence arbovirus emergence processes. This review focuses on concepts from disease ecology, explaining the interplay between biodiversity and pathogen emergence.
Collapse
Affiliation(s)
- Marco Marklewitz
- Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Germany; German Center for Infection Research (DZIF), Germany
| | - Sandra Junglen
- Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-University Berlin, and Berlin Institute of Health, Germany; German Center for Infection Research (DZIF), Germany.
| |
Collapse
|
120
|
Induction and Suppression of NF-κB Signalling by a DNA Virus of Drosophila. J Virol 2019; 93:JVI.01443-18. [PMID: 30404807 DOI: 10.1128/jvi.01443-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 12/12/2022] Open
Abstract
Interactions between the insect immune system and RNA viruses have been extensively studied in Drosophila, in which RNA interference, NF-κB, and JAK-STAT pathways underlie antiviral immunity. In response to RNA interference, insect viruses have convergently evolved suppressors of this pathway that act by diverse mechanisms to permit viral replication. However, interactions between the insect immune system and DNA viruses have received less attention, primarily because few Drosophila-infecting DNA virus isolates are available. In this study, we used a recently isolated DNA virus of Drosophila melanogaster, Kallithea virus (KV; family Nudiviridae), to probe known antiviral immune responses and virus evasion tactics in the context of DNA virus infection. We found that fly mutants for RNA interference and immune deficiency (Imd), but not Toll, pathways are more susceptible to Kallithea virus infection. We identified the Kallithea virus-encoded protein gp83 as a potent inhibitor of Toll signalling, suggesting that Toll mediates antiviral defense against Kallithea virus infection but that it is suppressed by the virus. We found that Kallithea virus gp83 inhibits Toll signalling through the regulation of NF-κB transcription factors. Furthermore, we found that gp83 of the closely related Drosophila innubila nudivirus (DiNV) suppresses D. melanogaster Toll signalling, suggesting an evolutionarily conserved function of Toll in defense against DNA viruses. Together, these results provide a broad description of known antiviral pathways in the context of DNA virus infection and identify the first Toll pathway inhibitor in a Drosophila virus, extending the known diversity of insect virus-encoded immune inhibitors.IMPORTANCE Coevolution of multicellular organisms and their natural viruses may lead to an intricate relationship in which host survival requires effective immunity and virus survival depends on evasion of such responses. Insect antiviral immunity and reciprocal virus immunosuppression tactics have been well studied in Drosophila melanogaster, primarily during RNA, but not DNA, virus infection. Therefore, we describe interactions between a recently isolated Drosophila DNA virus (Kallithea virus [KV]) and immune processes known to control RNA viruses, such as RNA interference (RNAi) and Imd pathways. We found that KV suppresses the Toll pathway and identified gp83 as a KV-encoded protein that underlies this suppression. This immunosuppressive ability is conserved in another nudivirus, suggesting that the Toll pathway has conserved antiviral activity against DNA nudiviruses, which have evolved suppressors in response. Together, these results indicate that DNA viruses induce and suppress NF-κB responses, and they advance the application of KV as a model to study insect immunity.
Collapse
|
121
|
Valanne S, Salminen TS, Järvelä-Stölting M, Vesala L, Rämet M. Immune-inducible non-coding RNA molecule lincRNA-IBIN connects immunity and metabolism in Drosophila melanogaster. PLoS Pathog 2019; 15:e1007504. [PMID: 30633769 PMCID: PMC6345493 DOI: 10.1371/journal.ppat.1007504] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 01/24/2019] [Accepted: 12/05/2018] [Indexed: 01/09/2023] Open
Abstract
Non-coding RNAs have important roles in regulating physiology, including immunity. Here, we performed transcriptome profiling of immune-responsive genes in Drosophila melanogaster during a Gram-positive bacterial infection, concentrating on long non-coding RNA (lncRNA) genes. The gene most highly induced by a Micrococcus luteus infection was CR44404, named Induced by Infection (lincRNA-IBIN). lincRNA-IBIN is induced by both Gram-positive and Gram-negative bacteria in Drosophila adults and parasitoid wasp Leptopilina boulardi in Drosophila larvae, as well as by the activation of the Toll or the Imd pathway in unchallenged flies. We show that upon infection, lincRNA-IBIN is expressed in the fat body, in hemocytes and in the gut, and its expression is regulated by NF-κB signaling and the chromatin modeling brahma complex. In the fat body, overexpression of lincRNA-IBIN affected the expression of Toll pathway -mediated genes. Notably, overexpression of lincRNA-IBIN in unchallenged flies elevated sugar levels in the hemolymph by enhancing the expression of genes important for glucose retrieval. These data show that lncRNA genes play a role in Drosophila immunity and indicate that lincRNA-IBIN acts as a link between innate immune responses and metabolism.
Collapse
Affiliation(s)
- Susanna Valanne
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Tiina S. Salminen
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mirva Järvelä-Stölting
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Laura Vesala
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech Institute and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- PEDEGO Research Unit, and Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- * E-mail:
| |
Collapse
|
122
|
Ho T, Panyim S, Udomkit A. Suppression of argonautes compromises viral infection in Penaeus monodon. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 90:130-137. [PMID: 30227218 DOI: 10.1016/j.dci.2018.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 06/08/2023]
Abstract
Argonaute (Ago) proteins, the catalytic component of an RNA-induced silencing complex (RISC) in RNA interference pathway, function in diverse processes, especially in antiviral defense and transposon regulation. So far, cDNAs encoding four members of Argonaute were found in Penaeus monodon (PmAgo1-4). Two PmAgo proteins, PmAgo1 and PmAgo3 shared high percentage of amino acid identity to Ago1 and Ago2, respectively in other Penaeid shrimps. Therefore, the possible roles of PmAgo1 and PmAgo3 upon viral infection in shrimp were characterized in this study. The level of PmAgo1 mRNA expression in shrimp hemolymph was stimulated upon YHV challenge, but not with dsRNA administration. Interestingly, silencing of either PmAgo1 or PmAgo3 using sequence-specific dsRNAs impaired the efficiency of PmRab7-dsRNA to knockdown shrimp endogenous PmRab7 expression. Inhibition of yellow head virus (YHV) replication and delayed mortality rate were also observed in both PmAgo1-and PmAgo3-knockdown shrimp. In addition, silencing of PmAgo3 transcript, but not PmAgo1, revealed partial inhibition of white spot syndrome virus (WSSV) infection and delayed mortality rate. Therefore, our study provides insights into PmAgo1and PmAgo3 functions that are involved in a dsRNA-mediated gene silencing pathway and play roles in YHV and WSSV replication in the shrimp.
Collapse
Affiliation(s)
- Teerapong Ho
- Institute of Molecular Biosciences, Mahidol University, Phutthamonthon 4 Road, Salaya, Nahkon Pathom, 73170, Thailand
| | - Sakol Panyim
- Institute of Molecular Biosciences, Mahidol University, Phutthamonthon 4 Road, Salaya, Nahkon Pathom, 73170, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Apinunt Udomkit
- Institute of Molecular Biosciences, Mahidol University, Phutthamonthon 4 Road, Salaya, Nahkon Pathom, 73170, Thailand.
| |
Collapse
|
123
|
Nucleic Acid Sensing in Invertebrate Antiviral Immunity. NUCLEIC ACID SENSING AND IMMUNITY - PART B 2019; 345:287-360. [DOI: 10.1016/bs.ircmb.2018.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
124
|
Drosophila as a Model to Study Brain Innate Immunity in Health and Disease. Int J Mol Sci 2018; 19:ijms19123922. [PMID: 30544507 PMCID: PMC6321579 DOI: 10.3390/ijms19123922] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022] Open
Abstract
Innate immunity is the first line of defense against invading pathogens and plays an essential role in defending the brain against infection, injury, and disease. It is currently well recognized that central nervous system (CNS) infections can result in long-lasting neurological sequelae and that innate immune and inflammatory reactions are highly implicated in the pathogenesis of neurodegeneration. Due to the conservation of the mechanisms that govern neural development and innate immune activation from flies to mammals, the lack of a classical adaptive immune system and the availability of numerous genetic and genomic tools, the fruit fly Drosophila melanogaster presents opportunities to investigate the cellular and molecular mechanisms associated with immune function in brain tissue and how they relate to infection, injury and neurodegenerative diseases. Here, we present an overview of currently identified innate immune mechanisms specific to the adult Drosophila brain.
Collapse
|
125
|
Donald CL, Varjak M, Aguiar ERGR, Marques JT, Sreenu VB, Schnettler E, Kohl A. Antiviral RNA Interference Activity in Cells of the Predatory Mosquito, Toxorhynchites amboinensis. Viruses 2018; 10:v10120694. [PMID: 30563205 PMCID: PMC6316411 DOI: 10.3390/v10120694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/19/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022] Open
Abstract
Arthropod vectors control the replication of arboviruses through their innate antiviral immune responses. In particular, the RNA interference (RNAi) pathways are of notable significance for the control of viral infections. Although much has been done to understand the role of RNAi in vector populations, little is known about its importance in non-vector mosquito species. In this study, we investigated the presence of an RNAi response in Toxorhynchites amboinensis, which is a non-blood feeding species proposed as a biological control agent against pest mosquitoes. Using a derived cell line (TRA-171), we demonstrate that these mosquitoes possess a functional RNAi response that is active against a mosquito-borne alphavirus, Semliki Forest virus. As observed in vector mosquito species, small RNAs are produced that target viral sequences. The size and characteristics of these small RNAs indicate that both the siRNA and piRNA pathways are induced in response to infection. Taken together, this data suggests that Tx. amboinensis are able to control viral infections in a similar way to natural arbovirus vector mosquito species. Understanding their ability to manage arboviral infections will be advantageous when assessing these and similar species as biological control agents.
Collapse
Affiliation(s)
- Claire L Donald
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland G61 1QH, UK.
| | - Margus Varjak
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland G61 1QH, UK.
| | - Eric Roberto Guimarães Rocha Aguiar
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha-Belo Horizonte-MG, CEP 31270-901, Brazil.
| | - João T Marques
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha-Belo Horizonte-MG, CEP 31270-901, Brazil.
| | - Vattipally B Sreenu
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland G61 1QH, UK.
| | - Esther Schnettler
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland G61 1QH, UK.
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland G61 1QH, UK.
| |
Collapse
|
126
|
Kariithi HM, Boucias DG, Murungi EK, Meki IK, Demirbaş-Uzel G, van Oers MM, Vreysen MJB, Abd-Alla AMM, Vlak JM. Coevolution of hytrosaviruses and host immune responses. BMC Microbiol 2018; 18:183. [PMID: 30470186 PMCID: PMC6251100 DOI: 10.1186/s12866-018-1296-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Hytrosaviruses (SGHVs; Hytrosaviridae family) are double-stranded DNA (dsDNA) viruses that cause salivary gland hypertrophy (SGH) syndrome in flies. Two structurally and functionally distinct SGHVs are recognized; Glossina pallidipes SGHV (GpSGHV) and Musca domestica SGHV (MdSGHV), that infect the hematophagous tsetse fly and the filth-feeding housefly, respectively. Genome sizes and gene contents of GpSGHV (~ 190 kb; 160-174 genes) and MdSGHV (~ 124 kb; 108 genes) may reflect an evolution with the SGHV-hosts resulting in differences in pathobiology. Whereas GpSGHV can switch from asymptomatic to symptomatic infections in response to certain unknown cues, MdSGHV solely infects symptomatically. Overt SGH characterizes the symptomatic infections of SGHVs, but whereas MdSGHV induces both nuclear and cellular hypertrophy (enlarged non-replicative cells), GpSGHV induces cellular hyperplasia (enlarged replicative cells). Compared to GpSGHV's specificity to Glossina species, MdSGHV infects other sympatric muscids. The MdSGHV-induced total shutdown of oogenesis inhibits its vertical transmission, while the GpSGHV's asymptomatic and symptomatic infections promote vertical and horizontal transmission, respectively. This paper reviews the coevolution of the SGHVs and their hosts (housefly and tsetse fly) based on phylogenetic relatedness of immune gene orthologs/paralogs and compares this with other virus-insect models. RESULTS Whereas MdSGHV is not vertically transmitted, GpSGHV is both vertically and horizontally transmitted, and the balance between the two transmission modes may significantly influence the pathogenesis of tsetse virus. The presence and absence of bacterial symbionts (Wigglesworthia and Sodalis) in tsetse and Wolbachia in the housefly, respectively, potentially contributes to the development of SGH symptoms. Unlike MdSGHV, GpSGHV contains not only host-derived proteins, but also appears to have evolutionarily recruited cellular genes from ancestral host(s) into its genome, which, although may be nonessential for viral replication, potentially contribute to the evasion of host's immune responses. Whereas MdSGHV has evolved strategies to counteract both the housefly's RNAi and apoptotic responses, the housefly has expanded its repertoire of immune effector, modulator and melanization genes compared to the tsetse fly. CONCLUSIONS The ecologies and life-histories of the housefly and tsetse fly may significantly influence coevolution of MdSGHV and GpSGHV with their hosts. Although there are still many unanswered questions regarding the pathogenesis of SGHVs, and the extent to which microbiota influence expression of overt SGH symptoms, SGHVs are attractive 'explorers' to elucidate the immune responses of their hosts, and the transmission modes of other large DNA viruses.
Collapse
Affiliation(s)
- Henry M Kariithi
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, Kaptagat Rd, Loresho, Nairobi, 00200, Kenya. .,Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Wagrammer Straße 5, A-1400, Vienna, Austria. .,Present Address: US National Poultry Research Centre, Southeast Poultry Research Laboratory, USDA-ARS, 934 College Station Road, Athens, GA, 30605, USA.
| | - Drion G Boucias
- Entomology and Nematology Department, University of Florida, 970 Natural Area Drive, Gainesville, FL, 32611, USA
| | - Edwin K Murungi
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Njoro, 20115, Kenya
| | - Irene K Meki
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Wagrammer Straße 5, A-1400, Vienna, Austria.,Laboratory of Virology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Güler Demirbaş-Uzel
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Wagrammer Straße 5, A-1400, Vienna, Austria
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Marc J B Vreysen
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Wagrammer Straße 5, A-1400, Vienna, Austria
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Wagrammer Straße 5, A-1400, Vienna, Austria
| | - Just M Vlak
- Laboratory of Virology, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| |
Collapse
|
127
|
Meki IK, Kariithi HM, Parker AG, Vreysen MJB, Ros VID, Vlak JM, van Oers MM, Abd-Alla AMM. RNA interference-based antiviral immune response against the salivary gland hypertrophy virus in Glossina pallidipes. BMC Microbiol 2018; 18:170. [PMID: 30470195 PMCID: PMC6251114 DOI: 10.1186/s12866-018-1298-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Glossina pallidipes salivary gland hypertrophy virus (GpSGHV; Hytrosaviridae) is a non-occluded dsDNA virus that specifically infects the adult stages of the hematophagous tsetse flies (Glossina species, Diptera: Glossinidae). GpSGHV infections are usually asymptomatic, but unknown factors can result to a switch to acute symptomatic infection, which is characterized by the salivary gland hypertrophy (SGH) syndrome associated with decreased fecundity that can ultimately lead to a colony collapse. It is uncertain how GpSGHV is maintained amongst Glossina spp. populations but RNA interference (RNAi) machinery, a conserved antiviral defense in insects, is hypothesized to be amongst the host’s mechanisms to maintain the GpSGHV in asymptomatic (persistent or latent) infection state. Here, we investigated the involvement of RNAi during GpSGHV infections by comparing the expression of three key RNAi machinery genes, Dicer (DCR), Argonaute (AGO) and Drosha, in artificially virus injected, asymptomatic and symptomatic infected G. pallidipes flies compared to PBS injected (controls) individuals. We further assessed the impact of AGO2 knockdown on virus infection by RT-qPCR quantification of four selected GpSGHV genes, i.e. odv-e66, dnapol, maltodextrin glycosyltransferase (a tegument gene) and SGHV091 (a capsid gene). Results We show that in response to hemocoelic injections of GpSGHV into G. pallidipes flies, increased virus replication was accompanied by significant upregulation of the expression of three RNAi key genes; AGO1, AGO2 and DCR2, and a moderate increase in the expression of Drosha post injection compared to the PBS-injected controls. Furthermore, compared to asymptomatically infected individuals, symptomatic flies showed significant downregulation of AGO1, AGO2 and Drosha, but a moderate increase in the expression of DCR2. Compared to the controls, knockdown of AGO2 did not have a significant impact on virus infection in the flies as evidenced by unaltered transcript levels of the selected GpSGHV genes. Conclusion The upregulation of the expression of the RNAi genes implicate involvement of this machinery in controlling GpSGHV infections and the establishment of symptomatic GpSGHV infections in Glossina. These findings provide a strategic foundation to understand GpSGHV infections and to control latent (asymptomatic) infections in Glossina spp. and thereby control SGHVs in insect production facilities. Electronic supplementary material The online version of this article (10.1186/s12866-018-1298-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irene K Meki
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria.,Laboratory of Virology, Wageningen University, 6708, PB, Wageningen, The Netherlands
| | - Henry M Kariithi
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria.,Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O Box 57811, Loresho, Nairobi, Kenya
| | - Andrew G Parker
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria
| | - Marc J B Vreysen
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria
| | - Vera I D Ros
- Laboratory of Virology, Wageningen University, 6708, PB, Wageningen, The Netherlands
| | - Just M Vlak
- Laboratory of Virology, Wageningen University, 6708, PB, Wageningen, The Netherlands
| | - Monique M van Oers
- Laboratory of Virology, Wageningen University, 6708, PB, Wageningen, The Netherlands
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria.
| |
Collapse
|
128
|
Regulating gene expression in animals through RNA endonucleolytic cleavage. Heliyon 2018; 4:e00908. [PMID: 30426105 PMCID: PMC6223193 DOI: 10.1016/j.heliyon.2018.e00908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/28/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The expression of any gene must be precisely controlled for appropriate function. This expression can be controlled at various levels. This includes epigenetic regulation through DNA methylation or histone modifications. At the posttranscriptional level, regulation can be via alternative splicing or controlling messenger RNA (mRNA) stability. RNA cleavage is one way to control mRNA stability. For example, microRNA (miRNA)-induced mRNA cleavage has long been recognised in plants. RNA cleavage also appears to be widespread in other kingdoms of life, and it is now clear that mRNA cleavage plays critical functions in animals. Although miRNA-induced mRNA cleavage can occur in animals, it is not a widespread mechanism. Instead, mRNA cleavage can be induced by a range of other mechanisms, including by endogenous short inhibitory RNAs (endo-siRNAs), as well as the Ribonuclease III (RNase III) enzymes Drosha and Dicer. In addition, RNA cleavage induced by endo-siRNAs and PIWI-interacting RNAs (piRNAs) is important for genome defence against transposons. Moreover, several RNase has been identified as important antiviral mediators. In this review, we will discuss these various RNA endonucleolytic cleavage mechanisms utilised by animals to regulate the expression of genes and as a defence against retrotransposons and viral infection.
Collapse
|
129
|
Jing T, Wang F, Qi F, Wang Z. Insect anal droplets contain diverse proteins related to gut homeostasis. BMC Genomics 2018; 19:784. [PMID: 30376807 PMCID: PMC6208037 DOI: 10.1186/s12864-018-5182-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/17/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Insects share similar fundamental molecular principles with mammals in innate immunity. For modulating normal gut microbiota, insects produce phenoloxidase (PO), which is absent in all vertebrates, and reactive nitrogen species (ROS) and antimicrobial proteins (AMPs). However, reports on insect gut phagocytosis are very few. Furthermore, most previous studies measure gene expression at the transcription level. In this study, we provided proteomic evidence on gut modulation of normal microorganisms by investigating the anal droplets from a weevil, Cryptorhynchus lapathi. RESULTS The results showed that the anal droplets contained diverse proteins related to physical barriers, epithelium renewal, pattern recognition, phenoloxidase activation, oxidative defense and phagocytosis, but AMPs were not detected. According to annotations, Scarb1, integrin βν, Dscam, spondin or Thbs2s might mediate phagocytosis. As a possible integrin βν pathway, βν activates Rho by an unknown mechanism, and Rho induces accumulation of mDia, which then promotes actin polymerization. CONCLUSIONS Our results well demonstrated that insect anal droplets can be used as materials to investigate the defense of a host to gut microorganisms and supported to the hypothesis that gut phagocytosis occurs in insects.
Collapse
Affiliation(s)
- Tianzhong Jing
- School of Forestry, Northeast Forestry University, Harbin, 150040, China.
| | - Fuxiao Wang
- School of Forestry, Northeast Forestry University, Harbin, 150040, China
| | - Fenghui Qi
- School of Life Sciences, Northeast Forestry University, Harbin, 150040, China
| | - Zhiying Wang
- School of Forestry, Northeast Forestry University, Harbin, 150040, China
| |
Collapse
|
130
|
Mondotte JA, Gausson V, Frangeul L, Blanc H, Lambrechts L, Saleh MC. Immune priming and clearance of orally acquired RNA viruses in Drosophila. Nat Microbiol 2018; 3:1394-1403. [PMID: 30374170 DOI: 10.1038/s41564-018-0265-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/06/2018] [Indexed: 12/17/2022]
Abstract
Immune responses in insects are differentially triggered depending on the infection route used by the pathogen. In most studies involving Drosophila melanogaster and viruses, infection is done by injection, while oral infection, which is probably the most common route of viral entry in nature, remains unexplored. Here, we orally infected adults and larvae from wild-type and RNA interference (RNAi) mutant flies with different RNA viruses. We found that, in contrast with what is observed following virus injection, oral infections initiated at larval or adult stages are cleared in adult flies. Virus elimination occurred despite a larger infectious dose than for injected flies and evidence of viral replication. RNAi mutant flies suffered greater mortality relative to wild-type flies following oral infection, but they also eliminated the virus, implying that RNAi is not essential for viral clearance and that other immune mechanisms act during oral infections. We further showed that information of infection by RNA viruses acquired orally leaves a trace under a DNA form, which confers protection against future reinfection by the same virus. Together, this work presents evidence of clearance and immune priming for RNA viruses in insects and challenges the current view of antiviral immunity in insects.
Collapse
Affiliation(s)
- Juan A Mondotte
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Valérie Gausson
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Lionel Frangeul
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions Group, Department of Genomes and Genetics, CNRS Unité Mixte de Recherche 2000, Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Department of Virology, CNRS Unité Mixte de Recherche 3569, Paris, France.
| |
Collapse
|
131
|
Abstract
There are several RNA interference (RNAi) pathways in insects. The small interfering RNA pathway is considered to be the main antiviral mechanism of the innate immune system; however, virus-specific P-element-induced Wimpy testis gene (PIWI)-interacting RNAs (vpiRNAs) have also been described, especially in mosquitoes. Understanding the antiviral potential of the RNAi pathways is important, given that many human and animal pathogens are transmitted by mosquitoes, such as Zika virus, dengue virus and chikungunya virus. In recent years, significant progress has been made to characterize the piRNA pathway in mosquitoes (including the possible antiviral activity) and to determine the differences between mosquitoes and the model organism Drosophila melanogaster. The new findings, especially regarding vpiRNA in mosquitoes, as well as important questions that need to be tackled in the future, are discussed in this review.
Collapse
Affiliation(s)
- Margus Varjak
- 1MRC - University of Glasgow Centre for Virus Research, Glasgow, G61 1QH, UK
| | - Mayke Leggewie
- 2Bernhard-Nocht-Institute for Tropical Medicine, Bernhard-Nocht-Strasse 74, 20359 Hamburg, Germany.,3German Centre for Infection Research (DZIF), partner site Hamburg-Luebeck-Borstel-Riems, Hamburg 20359, Germany
| | - Esther Schnettler
- 2Bernhard-Nocht-Institute for Tropical Medicine, Bernhard-Nocht-Strasse 74, 20359 Hamburg, Germany.,3German Centre for Infection Research (DZIF), partner site Hamburg-Luebeck-Borstel-Riems, Hamburg 20359, Germany
| |
Collapse
|
132
|
Nayak A, Kim DY, Trnka MJ, Kerr CH, Lidsky PV, Stanley DJ, Rivera BM, Li KH, Burlingame AL, Jan E, Frydman J, Gross JD, Andino R. A Viral Protein Restricts Drosophila RNAi Immunity by Regulating Argonaute Activity and Stability. Cell Host Microbe 2018; 24:542-557.e9. [PMID: 30308158 PMCID: PMC6450077 DOI: 10.1016/j.chom.2018.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 07/13/2018] [Accepted: 09/10/2018] [Indexed: 11/30/2022]
Abstract
The dicistrovirus, Cricket paralysis virus (CrPV) encodes an RNA interference (RNAi) suppressor, 1A, which modulates viral virulence. Using the Drosophila model, we combined structural, biochemical, and virological approaches to elucidate the strategies by which CrPV-1A restricts RNAi immunity. The atomic resolution structure of CrPV-1A uncovered a flexible loop that interacts with Argonaute 2 (Ago-2), thereby inhibiting Ago-2 endonuclease-dependent immunity. Mutations disrupting Ago-2 binding attenuates viral pathogenesis in wild-type but not Ago-2-deficient flies. CrPV-1A also contains a BC-box motif that enables the virus to hijack a host Cul2-Rbx1-EloBC ubiquitin ligase complex, which promotes Ago-2 degradation and virus replication. Our study uncovers a viral-based dual regulatory program that restricts antiviral immunity by direct interaction with and modulation of host proteins. While the direct inhibition of Ago-2 activity provides an efficient mechanism to establish infection, the recruitment of a ubiquitin ligase complex enables CrPV-1A to amplify Ago-2 inactivation to restrict further antiviral RNAi immunity.
Collapse
Affiliation(s)
- Arabinda Nayak
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA; Department of Biology and Genetics, Stanford University, Stanford, CA 94305, USA
| | - Dong Young Kim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA; College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 38541, South Korea
| | - Michael J Trnka
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Craig H Kerr
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Peter V Lidsky
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA
| | - David J Stanley
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Brianna Monique Rivera
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kathy H Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Eric Jan
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Judith Frydman
- Department of Biology and Genetics, Stanford University, Stanford, CA 94305, USA
| | - John D Gross
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
133
|
Harsh S, Ozakman Y, Kitchen SM, Paquin-Proulx D, Nixon DF, Eleftherianos I. Dicer-2 Regulates Resistance and Maintains Homeostasis against Zika Virus Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2018; 201:3058-3072. [PMID: 30305326 DOI: 10.4049/jimmunol.1800597] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) outbreaks pose a massive public health threat in several countries. We have developed an in vivo model to investigate the host-ZIKV interaction in Drosophila We have found that a strain of ZIKV replicates in wild-type flies without reducing their survival ability. We have shown that ZIKV infection triggers RNA interference and that mutating Dicer-2 results in enhanced ZIKV load and increased susceptibility to ZIKV infection. Using a flavivirus-specific Ab, we have found that ZIKV is localized in the gut and fat body cells of the infected wild-type flies and results in their perturbed homeostasis. In addition, Dicer-2 mutants display severely reduced insulin activity, which could contribute toward the increased mortality of these flies. Our work establishes the suitability of Drosophila as the model system to study host-ZIKV dynamics, which is expected to greatly advance our understanding of the molecular and physiological processes that determine the outcome of this disease.
Collapse
Affiliation(s)
- Sneh Harsh
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Yaprak Ozakman
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Shannon M Kitchen
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Dominic Paquin-Proulx
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Douglas F Nixon
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| |
Collapse
|
134
|
Yang M, Xu Z, Zhao W, Liu Q, Li Q, Lu L, Liu R, Zhang X, Cui F. Rice stripe virus-derived siRNAs play different regulatory roles in rice and in the insect vector Laodelphax striatellus. BMC PLANT BIOLOGY 2018; 18:219. [PMID: 30286719 PMCID: PMC6172784 DOI: 10.1186/s12870-018-1438-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/23/2018] [Indexed: 05/20/2023]
Abstract
BACKGROUND Most plant viruses depend on vector insects for transmission. Upon viral infection, virus-derived small interfering RNAs (vsiRNAs) can target both viral and host transcripts. Rice stripe virus (RSV) is a persistent-propagative virus transmitted by the small brown planthopper (Laodelphax striatellus, Fallen) and can cause a severe disease on rice. RESULTS To investigate how vsiRNAs regulate gene expressions in the host plant and the insect vector, we analyzed the expression profiles of small RNAs (sRNAs) and mRNAs in RSV-infected rice and RSV-infected planthopper. We obtained 88,247 vsiRNAs in rice that were predominantly derived from the terminal regions of the RSV RNA segments, and 351,655 vsiRNAs in planthopper that displayed relatively even distributions on RSV RNA segments. 38,112 and 80,698 unique vsiRNAs were found only in rice and planthopper, respectively, while 14,006 unique vsiRNAs were found in both of them. Compared to mock-inoculated rice, 273 genes were significantly down-regulated genes (DRGs) in RSV-infected rice, among which 192 (70.3%) were potential targets of vsiRNAs based on sequence complementarity. Gene ontology (GO) analysis revealed that these 192 DRGs were enriched in genes involved in kinase activity, carbohydrate binding and protein binding. Similarly, 265 DRGs were identified in RSV-infected planthoppers, among which 126 (47.5%) were potential targets of vsiRNAs. These planthopper target genes were enriched in genes that are involved in structural constituent of cuticle, serine-type endopeptidase activity, and oxidoreductase activity. CONCLUSIONS Taken together, our results reveal that infection by the same virus can generate distinct vsiRNAs in different hosts to potentially regulate different biological processes, thus reflecting distinct virus-host interactions.
Collapse
Affiliation(s)
- Meiling Yang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
| | - Zhongtian Xu
- Shanghai Center for Plant Stress Biology, Chinese Academy of Sciences, Shanghai, 201602 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Wan Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
| | - Qing Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Qiong Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Lu Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
| | - Renyi Liu
- Center for Agroforestry Mega Data Science and FAFU-UCR Joint Center for Horticultural Biology and Metabolomics, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, 350002 China
| | - Xiaoming Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
| | - Feng Cui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Bei Chen Xi Lu 1-5, Beijing, 100101 China
| |
Collapse
|
135
|
Fay EJ, Langlois RA. MicroRNA-Attenuated Virus Vaccines. Noncoding RNA 2018; 4:E25. [PMID: 30279330 PMCID: PMC6316615 DOI: 10.3390/ncrna4040025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 12/18/2022] Open
Abstract
Live-attenuated vaccines are the most effective way to establish robust, long-lasting immunity against viruses. However, the possibility of reversion to wild type replication and pathogenicity raises concerns over the safety of these vaccines. The use of host-derived microRNAs (miRNAs) to attenuate viruses has been accomplished in an array of biological contexts. The broad assortment of effective tissue- and species-specific miRNAs, and the ability to target a virus with multiple miRNAs, allow for targeting to be tailored to the virus of interest. While escape is always a concern, effective strategies have been developed to improve the safety and stability of miRNA-attenuated viruses. In this review, we discuss the various approaches that have been used to engineer miRNA-attenuated viruses, the steps that have been taken to improve their safety, and the potential use of these viruses as vaccines.
Collapse
Affiliation(s)
- Elizabeth J Fay
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ryan A Langlois
- Biochemistry, Molecular Biology, and Biophysics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
136
|
Karamipour N, Fathipour Y, Talebi AA, Asgari S, Mehrabadi M. Small interfering RNA pathway contributes to antiviral immunity in Spodoptera frugiperda (Sf9) cells following Autographa californica multiple nucleopolyhedrovirus infection. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 101:24-31. [PMID: 30075239 DOI: 10.1016/j.ibmb.2018.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/22/2018] [Accepted: 07/29/2018] [Indexed: 06/08/2023]
Abstract
Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is a well-known virus in the Baculoviridae family. Presence of the p35 gene in the AcMNPV genome as a suppressor of the short interfering RNA (siRNA) pathway is a strong reason for the importance of the siRNA pathway in the host cellular defense. Given that, here we explored the roles of Dicer-2 (Dcr2) and Argonaute 2 (Ago2) genes, key factors in the siRNA pathway in response to AcMNPV infection in Spodoptera frugiperda Sf9 cells. The results showed that the transcript levels of Dcr2 and Ago2 increased in response to AcMNPV infection particularly over 16 h post infection suggesting induction of the siRNA pathway. Reductions in the expression levels of Dcr2 and Ago2 by using specific dsRNAs in Sf9 cells modestly enhanced production of viral genomic DNA which indicated their role in the host antiviral defense. Using deep sequencing, our previous study showed a large number of small reads (siRNAs of ∼20 nucleotides) from AcMNPV-infected Sf9 cells that were mapped to some of the viral genes (hot spots). Down-regulation of Dcr2 in Sf9 cells resulted in enhanced expression levels of the selected virus hotspot genes (i.e. ORF-9 and ORF-148), while the transcript levels of virus cold spots (i.e. ORF-18 and ORF-25) with no or few siRNAs mapped to them did not change. Overexpression of AcMNPV p35 as a suppressor of RNAi and anti-apoptosis gene in Sf9 cells increased virus replication. Also, replication of mutant AcMNPV lacking the p35 gene was significantly increased in Sf9 cells with reduced transcript levels of Dcr2 and Ago2, highlighting the antiviral role of the siRNA pathway in Sf9 cells. Together, our results demonstrate that Dcr2 and Ago2 genes contribute in efficient antiviral response of Sf9 cells towards AcMNPV, and in turn, the AcMNPV p35 suppresses the siRNA pathway, besides being an antiapoptotic protein.
Collapse
Affiliation(s)
- Naeime Karamipour
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathipour
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Ali Asghar Talebi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran
| | - Sassan Asgari
- Australian Infectious Disease Research Centre, School of Biological Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Mohammad Mehrabadi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
137
|
Abstract
Like humans, insects face the threat of viral infection. Despite having repercussions on human health and disease, knowledge gaps exist for how insects cope with viral pathogens. Drosophila melanogaster serves as an ideal insect model due to its genetic tractability. When encountering a pathogen, two major approaches to fight disease are resistance strategies and tolerance strategies. Disease resistance strategies promote the health of the infected host by reducing pathogen load. Multiple disease resistance mechanisms have been identified in Drosophila: RNA interference, Jak/STAT signaling, Toll signaling, IMD signaling, and autophagy. Disease tolerance mechanisms, in contrast, do not reduce pathogen load directly, but rather mitigate the stress and damage incurred by infection. The main benefit of tolerance mechanisms may therefore be to provide the host with time to engage antiviral resistance mechanisms that eliminate the threat. In this review, antiviral resistance mechanisms used by Drosophila will be described and compared to mammalian antiviral mechanisms. Disease tolerance will then be explained in a broader context as this is a burgeoning field of study.
Collapse
Affiliation(s)
- Jonathan Chow
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jonathan C Kagan
- Division of Gastroenterology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States.
| |
Collapse
|
138
|
Yang D, Xu X, Zhao H, Yang S, Wang X, Zhao D, Diao Q, Hou C. Diverse Factors Affecting Efficiency of RNAi in Honey Bee Viruses. Front Genet 2018; 9:384. [PMID: 30254665 PMCID: PMC6141667 DOI: 10.3389/fgene.2018.00384] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/27/2018] [Indexed: 12/15/2022] Open
Abstract
Infection and transmission of honey bee viruses pose a serious threat to the pollination services of crops and wild plants, which plays a vital role in agricultural economy and ecology. RNA interference (RNAi) is an effective defense mechanism against commonly occurring viral infections of animals and plants. However, recent studies indicate that the effects of RNAi on the honey bee can induce additional impacts and might not always be effective in suppressing the virus. Moreover, the RNAi responses differed in relation to the developmental stage of the insect and the target tissue used, even though the same method of delivery was used. These results indicate that further analysis and field experiments should be performed to characterize the varying effectiveness of RNAi-based methods for treating honey bee viral infections. In this review, we provide an overview of the current knowledge and the recent progress in RNAi-based anti-viral treatments for honey bees, focusing in particular highlight the role of the dsRNA-delivery method used and its effect on RNAi efficiency and demonstrate the potential practical value of this tool for controlling the virus. We conclude studying the gene function and disease control of honey bee by RNAi technology requires a complex consideration from physiology, genetics to environment.
Collapse
Affiliation(s)
- Dahe Yang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiang Xu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| | - Hongxia Zhao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangdong Institute of Applied Biological Resources, Guangzhou, China
| | - Sa Yang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| | - Xinling Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| | - Di Zhao
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| | - Qingyun Diao
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| | - Chunsheng Hou
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Beijing, China
| |
Collapse
|
139
|
Goto A, Okado K, Martins N, Cai H, Barbier V, Lamiable O, Troxler L, Santiago E, Kuhn L, Paik D, Silverman N, Holleufer A, Hartmann R, Liu J, Peng T, Hoffmann JA, Meignin C, Daeffler L, Imler JL. The Kinase IKKβ Regulates a STING- and NF-κB-Dependent Antiviral Response Pathway in Drosophila. Immunity 2018; 49:225-234.e4. [PMID: 30119996 DOI: 10.1016/j.immuni.2018.07.013] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/08/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022]
Abstract
Antiviral immunity in Drosophila involves RNA interference and poorly characterized inducible responses. Here, we showed that two components of the IMD pathway, the kinase dIKKβ and the transcription factor Relish, were required to control infection by two picorna-like viruses. We identified a set of genes induced by viral infection and regulated by dIKKβ and Relish, which included an ortholog of STING. We showed that dSTING participated in the control of infection by picorna-like viruses, acting upstream of dIKKβ to regulate expression of Nazo, an antiviral factor. Our data reveal an antiviral function for STING in an animal model devoid of interferons and suggest an evolutionarily ancient role for this molecule in antiviral immunity.
Collapse
Affiliation(s)
- Akira Goto
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| | - Kiyoshi Okado
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Nelson Martins
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Hua Cai
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Vincent Barbier
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Olivier Lamiable
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Laurent Troxler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Estelle Santiago
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Lauriane Kuhn
- Université de Strasbourg, CNRS, Plateforme Protéomique Strasbourg-Esplanade, 67000 Strasbourg, France
| | - Donggi Paik
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neal Silverman
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Andreas Holleufer
- Center for Structural Biology, Aarhus University, 8000 Aarhus C, Denmark
| | - Rune Hartmann
- Center for Structural Biology, Aarhus University, 8000 Aarhus C, Denmark
| | - Jiyong Liu
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Tao Peng
- Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Jules A Hoffmann
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China; University of Strasbourg Institute for Advanced Study, 67000 Strasbourg, France
| | - Carine Meignin
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Laurent Daeffler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France
| | - Jean-Luc Imler
- Université de Strasbourg, CNRS, Insect Models of Innate Immunity (M3I; UPR9022), 67084 Strasbourg, France; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
140
|
McMenamin AJ, Daughenbaugh KF, Parekh F, Pizzorno MC, Flenniken ML. Honey Bee and Bumble Bee Antiviral Defense. Viruses 2018; 10:E395. [PMID: 30060518 PMCID: PMC6115922 DOI: 10.3390/v10080395] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
Bees are important plant pollinators in both natural and agricultural ecosystems. Managed and wild bees have experienced high average annual colony losses, population declines, and local extinctions in many geographic regions. Multiple factors, including virus infections, impact bee health and longevity. The majority of bee-infecting viruses are positive-sense single-stranded RNA viruses. Bee-infecting viruses often cause asymptomatic infections but may also cause paralysis, deformity or death. The severity of infection is governed by bee host immune responses and influenced by additional biotic and abiotic factors. Herein, we highlight studies that have contributed to the current understanding of antiviral defense in bees, including the Western honey bee (Apis mellifera), the Eastern honey bee (Apis cerana) and bumble bee species (Bombus spp.). Bee antiviral defense mechanisms include RNA interference (RNAi), endocytosis, melanization, encapsulation, autophagy and conserved immune pathways including Jak/STAT (Janus kinase/signal transducer and activator of transcription), JNK (c-Jun N-terminal kinase), MAPK (mitogen-activated protein kinases) and the NF-κB mediated Toll and Imd (immune deficiency) pathways. Studies in Dipteran insects, including the model organism Drosophila melanogaster and pathogen-transmitting mosquitos, provide the framework for understanding bee antiviral defense. However, there are notable differences such as the more prominent role of a non-sequence specific, dsRNA-triggered, virus limiting response in honey bees and bumble bees. This virus-limiting response in bees is akin to pathways in a range of organisms including other invertebrates (i.e., oysters, shrimp and sand flies), as well as the mammalian interferon response. Current and future research aimed at elucidating bee antiviral defense mechanisms may lead to development of strategies that mitigate bee losses, while expanding our understanding of insect antiviral defense and the potential evolutionary relationship between sociality and immune function.
Collapse
Affiliation(s)
- Alexander J McMenamin
- Department of Plant Sciences and Plant Pathology, Bozeman, MT 59717, USA.
- Department of Microbiology and Immunology, Bozeman, MT 59717, USA.
- Center for Pollinator Health, Montana State University, Bozeman, MT 59717, USA.
| | - Katie F Daughenbaugh
- Department of Plant Sciences and Plant Pathology, Bozeman, MT 59717, USA.
- Center for Pollinator Health, Montana State University, Bozeman, MT 59717, USA.
| | - Fenali Parekh
- Department of Plant Sciences and Plant Pathology, Bozeman, MT 59717, USA.
- Department of Microbiology and Immunology, Bozeman, MT 59717, USA.
- Center for Pollinator Health, Montana State University, Bozeman, MT 59717, USA.
| | - Marie C Pizzorno
- Biology Department, Bucknell University, Lewisburg, PA 17837, USA.
| | - Michelle L Flenniken
- Department of Plant Sciences and Plant Pathology, Bozeman, MT 59717, USA.
- Department of Microbiology and Immunology, Bozeman, MT 59717, USA.
- Center for Pollinator Health, Montana State University, Bozeman, MT 59717, USA.
| |
Collapse
|
141
|
Berkhout B. RNAi-mediated antiviral immunity in mammals. Curr Opin Virol 2018; 32:9-14. [PMID: 30015014 DOI: 10.1016/j.coviro.2018.07.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/26/2018] [Accepted: 07/08/2018] [Indexed: 12/18/2022]
Abstract
RNA interference (RNAi) was discovered in plants where it functions as the main antiviral pathway and this antiviral role was subsequently extended to invertebrates. But it remained hotly debated whether RNAi fulfils a similar role in mammals that already have a potent innate immune system based on interferon and an elaborate adaptive immune system. On the one hand, mammalian cells do encode most of the RNAi machinery, but this could be used exclusively to control cellular gene expression via micro RNAs (miRNAs). But on the other hand, virus-derived small interfering RNAs, the hallmark of RNAi involvement, could not be readily detected upon virus infection of mammalian cells. However, recent studies have indicated that these signature molecules are generated in virus-infected embryonic cell types of mammals and that viruses actively suppress such responses by means of potent RNAi suppressor proteins. Thus, the tide seems to be changing in favor of RNAi as accessory antiviral defense mechanism in humans. Intriguingly, recent studies indicate that insects have also developed an additional innate immune system that collaborates with the RNAi response in the fight against invading viral pathogens. Thus, the presence of multiple antiviral response mechanisms seems standard outside the plant world and we will specifically discuss the interactions between these antiviral programs.
Collapse
Affiliation(s)
- Ben Berkhout
- Laboratory of Experimental Virology, Department of Medical Microbiology, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
142
|
Yu R, Jing X, Li W, Xu J, Xu Y, Geng L, Zhu C, Liu H. Non-structural protein 1 from avian influenza virus H9N2 is an efficient RNA silencing suppressor with characteristics that differ from those of Tomato bushy stunt virus p19. Virus Genes 2018; 54:368-375. [PMID: 29480423 DOI: 10.1007/s11262-018-1544-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 02/19/2018] [Indexed: 10/17/2022]
Abstract
Non-structural protein 1 (NS1) of influenza A virus is a multifunctional dimeric protein that contains a conserved N-terminal RNA binding domain. Studies have shown that NS1 suppresses RNA silencing and the NS1 proteins encoded by different influenza A virus strains exhibit differential RNA silencing suppression activities. In this study, we showed that the NS1 protein from avian influenza virus (AIV) H9N2 suppressed systemic RNA silencing induced by sense RNA or dsRNA. It resulted in more severe Potato virus X symptom, but could not reverse established systemic green fluorescent protein silencing in Nicotiana benthamiana. In addition, its systemic silencing suppression activity was much weaker than that of p19. The local silencing suppression activity of AIV H9N2 NS1 was most powerful at 7 dpi and was even stronger than that of p19. And the inhibition ability to RNA silencing of NS1 is stronger than that of p19 in human cells. Collectively, these results indicate that AIV H9N2 NS1 is an effective RNA silencing suppressor that likely targets downstream step(s) of dsRNA formation at an early stage in RNA silencing. Although NS1 and p19 both bind siRNA, their suppression mechanisms seem to differ because of differences in their suppression activities at various times post-infiltration and because p19 can reverse established systemic RNA silencing, but NS1 cannot.
Collapse
Affiliation(s)
- Ru Yu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Xiuli Jing
- Institute of Immunology, Taishan Medical University, Tai'an, 271000, China
| | - Wenjing Li
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Jie Xu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Yang Xu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Liwei Geng
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Changxiang Zhu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Hongmei Liu
- State Key Laboratory of Crop Biology, College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
143
|
Camargo C, Wu K, Fishilevich E, Narva KE, Siegfried BD. Knockdown of RNA interference pathway genes in western corn rootworm, Diabrotica virgifera virgifera, identifies no fitness costs associated with Argonaute 2 or Dicer-2. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2018; 148:103-110. [PMID: 29891360 DOI: 10.1016/j.pestbp.2018.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/07/2018] [Accepted: 04/07/2018] [Indexed: 06/08/2023]
Abstract
The use of transgenic crops that induce silencing of essential genes using double-stranded RNA (dsRNA) through RNA interference (RNAi) in western corn rootworm, Diabrotica virgifera virgifera, is likely to be an important component of new technologies for the control of this important corn pest. Previous studies have demonstrated that the dsRNA response in D. v. virgifera depends on the presence of RNAi pathway genes including Dicer-2 and Argonaute 2, and that downregulation of these genes limits the lethality of environmental dsRNA. A potential resistance mechanism to lethal dsRNA may involve loss of function of RNAi pathway genes. Howver, the potential for resistance to evolve may depend on whether these pathway genes have essential functions such that the loss of function of core proteins in the RNAi pathway will have fitness costs in D. v. virgifera. Fitness costs associated with potential resistance mechanisms have a central role in determining how resistance can evolve to RNAi technologies in western corn rootworm. We evaluated the effect of dsRNA and microRNA pathway gene knockdown on the development of D. v. virgifera larvae through short-term and long-term exposures to dsRNA for Dicer and Argonaute genes. Downregulation of Argonaute 2, Dicer-2, Dicer-1 did not significantly affect larval survivorship or development through short and long-term exposure to dsRNA. However, downregulation of Argonaute 1 reduced larval survivorship and delayed development. The implications of these results as they relate to D. v. virgifera resistance to lethal dsRNA are discussed.
Collapse
Affiliation(s)
- Carolina Camargo
- Department of Entomology and Nematology, University of Florida, 1881 Natural Area Drive, Steinmetz Hall, Gainesville, FL 32611, United States
| | - Ke Wu
- Department of Entomology and Nematology, University of Florida, 1881 Natural Area Drive, Steinmetz Hall, Gainesville, FL 32611, United States
| | - Elane Fishilevich
- Dow AgroSciences, 9330 Zionsville Road, Indianapolis, IN 46268, United States
| | - Kenneth E Narva
- Dow AgroSciences, 9330 Zionsville Road, Indianapolis, IN 46268, United States
| | - Blair D Siegfried
- Department of Entomology and Nematology, University of Florida, 1881 Natural Area Drive, Steinmetz Hall, Gainesville, FL 32611, United States.
| |
Collapse
|
144
|
Liu T, Xu Y, Wang X, Gu J, Yan G, Chen XG. Antiviral systems in vector mosquitoes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:34-43. [PMID: 29294302 DOI: 10.1016/j.dci.2017.12.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/20/2017] [Accepted: 12/29/2017] [Indexed: 06/07/2023]
Abstract
Mosquito-borne viral diseases represent a major challenge to human public health. As natural vectors of arboviruses, mosquitoes can be infected by a virus, but they have evolved multiple mechanisms to tolerate constant infection and restrict viral replication via their antiviral immune system. In a state of continuous infection, a mosquito can transmit an arbovirus while obtaining a blood meal from a mammalian host. During infection, the virus is mainly inhibited through a small RNA-mediated interference mechanism. Within mosquitoes, the invaded viruses are recognized based on pathogen-associated molecular patterns, leading to the production of cytokines. These cytokines in turn bind pattern recognition receptors and activate Toll, IMD and other immune signalling pathways to expand the immune response and induce antiviral activity via immune effectors. Interestingly, the gut microbiota and Wolbachia also play a role in mosquito antiviral immunity, which is very similar to acquired immunity. This review describes the advances made in understanding various aspects of mosquito antiviral immune molecular mechanisms in detail and explores some of the unresolved issues related to the mosquito immune system.
Collapse
Affiliation(s)
- Tong Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Ye Xu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiaoming Wang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jinbao Gu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guiyun Yan
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China; Program in Public Health, School of Medicine, University of California, Irvine, USA
| | - Xiao-Guang Chen
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China.
| |
Collapse
|
145
|
McMahon DP, Wilfert L, Paxton RJ, Brown MJF. Emerging Viruses in Bees: From Molecules to Ecology. Adv Virus Res 2018; 101:251-291. [PMID: 29908591 DOI: 10.1016/bs.aivir.2018.02.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Emerging infectious diseases arise as a result of novel interactions between populations of hosts and pathogens, and can threaten the health and wellbeing of the entire spectrum of biodiversity. Bees and their viruses are a case in point. However, detailed knowledge of the ecological factors and evolutionary forces that drive disease emergence in bees and other host-pathogen communities is surprisingly lacking. In this review, we build on the fundamental insight that viruses evolve and adapt over timescales that overlap with host ecology. At the same time, we integrate the role of host community ecology, including community structure and composition, biodiversity loss, and human-driven disturbance, all of which represent significant factors in bee virus ecology. Both of these evolutionary and ecological perspectives represent major advances but, in most cases, it remains unclear how evolutionary forces actually operate across different biological scales (e.g., from cell to ecosystem). We present a molecule-to-ecology framework to help address these issues, emphasizing the role of molecular mechanisms as key bottom-up drivers of change at higher ecological scales. We consider the bee-virus system to be an ideal one in which to apply this framework. Unlike many other animal models, bees constitute a well characterized and accessible multispecies assemblage, whose populations and interspecific interactions can be experimentally manipulated and monitored in high resolution across space and time to provide robust tests of prevailing theory.
Collapse
Affiliation(s)
- Dino P McMahon
- Institute of Biology, Freie Universität Berlin, Berlin, Germany; Department for Materials and Environment, BAM Federal Institute for Materials Research and Testing, Berlin, Germany.
| | - Lena Wilfert
- Centre for Ecology and Conservation, University of Exeter, Penryn, United Kingdom
| | - Robert J Paxton
- Institute for Biology, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany; German Centre for integrative Biodiversity Research (iDiv), Halle-Jena-Leipzig, Leipzig, Germany
| | - Mark J F Brown
- School of Biological Sciences, Royal Holloway University of London, Egham, United Kingdom
| |
Collapse
|
146
|
Swevers L, Liu J, Smagghe G. Defense Mechanisms against Viral Infection in Drosophila: RNAi and Non-RNAi. Viruses 2018; 10:E230. [PMID: 29723993 PMCID: PMC5977223 DOI: 10.3390/v10050230] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/20/2018] [Accepted: 04/27/2018] [Indexed: 12/20/2022] Open
Abstract
RNAi is considered a major antiviral defense mechanism in insects, but its relative importance as compared to other antiviral pathways has not been evaluated comprehensively. Here, it is attempted to give an overview of the antiviral defense mechanisms in Drosophila that involve both RNAi and non-RNAi. While RNAi is considered important in most viral infections, many other pathways can exist that confer antiviral resistance. It is noted that very few direct recognition mechanisms of virus infections have been identified in Drosophila and that the activation of immune pathways may be accomplished indirectly through cell damage incurred by viral replication. In several cases, protection against viral infection can be obtained in RNAi mutants by non-RNAi mechanisms, confirming the variability of the RNAi defense mechanism according to the type of infection and the physiological status of the host. This analysis is aimed at more systematically investigating the relative contribution of RNAi in the antiviral response and more specifically, to ask whether RNAi efficiency is affected when other defense mechanisms predominate. While Drosophila can function as a useful model, this issue may be more critical for economically important insects that are either controlled (agricultural pests and vectors of diseases) or protected from parasite infection (beneficial insects as bees) by RNAi products.
Collapse
Affiliation(s)
- Luc Swevers
- Institute of Biosciences & Applications, NCSR "Demokritos", 15341 Athens, Greece.
| | - Jisheng Liu
- School of Life Sciences, Guangzhou University, 510006 Guangzhou, China.
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
147
|
Host shifts result in parallel genetic changes when viruses evolve in closely related species. PLoS Pathog 2018; 14:e1006951. [PMID: 29649296 PMCID: PMC5897010 DOI: 10.1371/journal.ppat.1006951] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/27/2018] [Indexed: 01/23/2023] Open
Abstract
Host shifts, where a pathogen invades and establishes in a new host species, are a major source of emerging infectious diseases. They frequently occur between related host species and often rely on the pathogen evolving adaptations that increase their fitness in the novel host species. To investigate genetic changes in novel hosts, we experimentally evolved replicate lineages of an RNA virus (Drosophila C Virus) in 19 different species of Drosophilidae and deep sequenced the viral genomes. We found a strong pattern of parallel evolution, where viral lineages from the same host were genetically more similar to each other than to lineages from other host species. When we compared viruses that had evolved in different host species, we found that parallel genetic changes were more likely to occur if the two host species were closely related. This suggests that when a virus adapts to one host it might also become better adapted to closely related host species. This may explain in part why host shifts tend to occur between related species, and may mean that when a new pathogen appears in a given species, closely related species may become vulnerable to the new disease. Host shifts, where a pathogen jumps from one host species to another, are a major source of infectious disease. Hosts shifts are more likely to occur between related host species and often rely on the pathogen evolving adaptations that increase their fitness in the novel host. Here we have investigated how viruses evolve in different host species, by experimentally evolving replicate lineages of an RNA virus in 19 different host species that shared a common ancestor 40 million years ago. We then deep sequenced the genomes of these viruses to examine the genetic changes that have occurred in different host species that vary in their relatedness. We found that parallel mutations–that are indicative of selection–were significantly more likely to occur within viral lineages from the same host, and between viruses evolved in closely related species. This suggests that a mutation that may adapt a virus to a given host, may also adapt it to closely related host species.
Collapse
|
148
|
Abstract
The power and ease of Drosophila genetics and the medical relevance of mosquito-transmitted viruses have made dipterans important model organisms in antiviral immunology. Studies of virus-host interactions at the molecular and population levels have illuminated determinants of resistance to virus infection. Here, we review the sources and nature of variation in antiviral immunity and virus susceptibility in model dipteran insects, specifically the fruit fly Drosophila melanogaster and vector mosquitoes of the genera Aedes and Culex. We first discuss antiviral immune mechanisms and describe the virus-specificity of these responses. In the following sections, we review genetic and microbiota-dependent variation in antiviral immunity. In the final sections, we explore less well-studied sources of variation, including abiotic factors, sexual dimorphism, infection history, and endogenous viral elements. We borrow from work on other pathogen types and non-dipteran species when it parallels or complements studies in dipterans. Understanding natural variation in virus-host interactions may lead to the identification of novel restriction factors and immune mechanisms and shed light on the molecular determinants of vector competence.
Collapse
Affiliation(s)
- William H Palmer
- Institute of Evolutionary Biology and Centre for Infection, Evolution and Immunity, University of Edinburgh, Edinburgh EH9 3FL UK.
| | - Finny S Varghese
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| | - Ronald P van Rij
- Department of Medical Microbiology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, P.O. Box 9101, Nijmegen 6500 HB, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen 6525 GA, The Netherlands.
| |
Collapse
|
149
|
Poirier EZ, Goic B, Tomé-Poderti L, Frangeul L, Boussier J, Gausson V, Blanc H, Vallet T, Loyd H, Levi LI, Lanciano S, Baron C, Merkling SH, Lambrechts L, Mirouze M, Carpenter S, Vignuzzi M, Saleh MC. Dicer-2-Dependent Generation of Viral DNA from Defective Genomes of RNA Viruses Modulates Antiviral Immunity in Insects. Cell Host Microbe 2018; 23:353-365.e8. [PMID: 29503180 PMCID: PMC5857290 DOI: 10.1016/j.chom.2018.02.001] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 11/08/2017] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
Abstract
The RNAi pathway confers antiviral immunity in insects. Virus-specific siRNA responses are amplified via the reverse transcription of viral RNA to viral DNA (vDNA). The nature, biogenesis, and regulation of vDNA are unclear. We find that vDNA produced during RNA virus infection of Drosophila and mosquitoes is present in both linear and circular forms. Circular vDNA (cvDNA) is sufficient to produce siRNAs that confer partially protective immunity when challenged with a cognate virus. cvDNAs bear homology to defective viral genomes (DVGs), and DVGs serve as templates for vDNA and cvDNA synthesis. Accordingly, DVGs promote the amplification of vDNA-mediated antiviral RNAi responses in infected Drosophila. Furthermore, vDNA synthesis is regulated by the DExD/H helicase domain of Dicer-2 in a mechanism distinct from its role in siRNA generation. We suggest that, analogous to mammalian RIG-I-like receptors, Dicer-2 functions like a pattern recognition receptor for DVGs to modulate antiviral immunity in insects.
Collapse
Affiliation(s)
- Enzo Z Poirier
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France; Institut Pasteur, Viral Populations and Pathogenesis, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France; University of Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, 75013 Paris, France
| | - Bertsy Goic
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Lorena Tomé-Poderti
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Lionel Frangeul
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Jérémy Boussier
- Institut Pasteur, Immunobiology of Dendritic Cells, Institut National de la Santé et de la Recherche Médicale, U1223, 75015 Paris, France
| | - Valérie Gausson
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France; Institut Pasteur, Viral Populations and Pathogenesis, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Hyelee Loyd
- Department of Animal Science, Iowa State University, Ames, IA 50010, USA
| | - Laura I Levi
- Institut Pasteur, Viral Populations and Pathogenesis, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Sophie Lanciano
- Institut de Recherche pour le Développement, DIADE, Université de Montpellier, Université de Perpignan, LGDP, 66860 Perpignan, France
| | - Chloé Baron
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France
| | - Sarah H Merkling
- Institut Pasteur, Insect-Virus Interactions, Centre National de la Recherche Scientifique URA 3012, 75015 Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Insect-Virus Interactions, Centre National de la Recherche Scientifique URA 3012, 75015 Paris, France
| | - Marie Mirouze
- Institut de Recherche pour le Développement, DIADE, Université de Montpellier, Université de Perpignan, LGDP, 66860 Perpignan, France
| | - Susan Carpenter
- Department of Animal Science, Iowa State University, Ames, IA 50010, USA
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France.
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference, Centre National de la Recherche Scientifique UMR 3569, 75015 Paris, France.
| |
Collapse
|
150
|
Spindle-E Acts Antivirally Against Alphaviruses in Mosquito Cells. Viruses 2018; 10:v10020088. [PMID: 29463033 PMCID: PMC5850395 DOI: 10.3390/v10020088] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022] Open
Abstract
Mosquitoes transmit several human- and animal-pathogenic alphaviruses (Togaviridae family). In alphavirus-infected mosquito cells two different types of virus-specific small RNAs are produced as part of the RNA interference response: short-interfering (si)RNAs and PIWI-interacting (pi)RNAs. The siRNA pathway is generally thought to be the main antiviral pathway. Although an antiviral activity has been suggested for the piRNA pathway its role in host defences is not clear. Knock down of key proteins of the piRNA pathway (Ago3 and Piwi5) in Aedes aegypti-derived cells reduced the production of alphavirus chikungunya virus (CHIKV)-specific piRNAs but had no effect on virus replication. In contrast, knock down of the siRNA pathway key protein Ago2 resulted in an increase in virus replication. Similar results were obtained when expression of Piwi4 was silenced. Knock down of the helicase Spindle-E (SpnE), an essential co-factor of the piRNA pathway in Drosophila melanogaster, resulted in increased virus replication indicating that SpnE acts as an antiviral against alphaviruses such as CHIKV and the related Semliki Forest virus (SFV). Surprisingly, this effect was found to be independent of the siRNA and piRNA pathways in Ae. aegypti cells and specific for alphaviruses. This suggests a small RNA-independent antiviral function for this protein in mosquitoes.
Collapse
|